Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(10): E1866-E1874, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223521

ABSTRACT

Here, we report a method for time-resolved, longitudinal extraction and quantitative measurement of intracellular proteins and mRNA from a variety of cell types. Cytosolic contents were repeatedly sampled from the same cell or population of cells for more than 5 d through a cell-culture substrate, incorporating hollow 150-nm-diameter nanostraws (NS) within a defined sampling region. Once extracted, the cellular contents were analyzed with conventional methods, including fluorescence, enzymatic assays (ELISA), and quantitative real-time PCR. This process was nondestructive with >95% cell viability after sampling, enabling long-term analysis. It is important to note that the measured quantities from the cell extract were found to constitute a statistically significant representation of the actual contents within the cells. Of 48 mRNA sequences analyzed from a population of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs), 41 were accurately quantified. The NS platform samples from a select subpopulation of cells within a larger culture, allowing native cell-to-cell contact and communication even during vigorous activity such as cardiomyocyte beating. This platform was applied both to cell lines and to primary cells, including CHO cells, hiPSC-CMs, and human astrocytes derived in 3D cortical spheroids. By tracking the same cell or group of cells over time, this method offers an avenue to understand dynamic cell behavior, including processes such as induced pluripotency and differentiation.


Subject(s)
Cell Tracking/methods , Embryonic Stem Cells/chemistry , Proteins/isolation & purification , RNA, Messenger/isolation & purification , Animals , CHO Cells , Cell Differentiation/drug effects , Cricetulus , Cytoplasm/chemistry , Cytoplasm/drug effects , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/cytology , Proteins/chemistry , RNA, Messenger/chemistry
2.
PLoS One ; 10(4): e0120093, 2015.
Article in English | MEDLINE | ID: mdl-25897839

ABSTRACT

The dynamic assembly of filamentous (F) actin plays essential roles in the assembly of presynaptic boutons, the fusion, mobilization and recycling of synaptic vesicles (SVs), and presynaptic forms of plasticity. However, the molecular mechanisms that regulate the temporal and spatial assembly of presynaptic F-actin remain largely unknown. Similar to other F-actin rich membrane specializations, presynaptic boutons contain a set of molecules that respond to cellular cues and trans-synaptic signals to facilitate activity-dependent assembly of F-actin. The presynaptic active zone (AZ) protein Piccolo has recently been identified as a key regulator of neurotransmitter release during SV cycling. It does so by coordinating the activity-dependent assembly of F-Actin and the dynamics of key plasticity molecules including Synapsin1, Profilin and CaMKII. The multidomain structure of Piccolo, its exquisite association with the AZ, and its ability to interact with a number of actin-associated proteins suggest that Piccolo may function as a platform to coordinate the spatial assembly of F-actin. Here we have identified Daam1, a Formin that functions with Profilin to drive F-actin assembly, as a novel Piccolo binding partner. We also found that within cells Daam1 activation promotes Piccolo binding, an interaction that can spatially direct the polymerization of F-Actin. Moreover, similar to Piccolo and Profilin, Daam1 loss of function impairs presynaptic-F-actin assembly in neurons. These data suggest a model in which Piccolo directs the assembly of presynaptic F-Actin from the AZ by scaffolding key actin regulatory proteins including Daam1.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/physiology , Presynaptic Terminals/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoskeletal Proteins/chemistry , Female , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Neuropeptides/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Pseudopodia/metabolism , Rats, Sprague-Dawley , Stress Fibers/metabolism
3.
EMBO J ; 32(7): 954-69, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23403927

ABSTRACT

The presynaptic active zone (AZ) is a specialized microdomain designed for the efficient and repetitive release of neurotransmitter. Bassoon and Piccolo are two high molecular weight components of the AZ, with hypothesized roles in its assembly and structural maintenance. However, glutamatergic synapses lacking either protein exhibit relatively minor defects, presumably due to their significant functional redundancy. In the present study, we have used interference RNAs to eliminate both proteins from glutamatergic synapses, and find that they are essential for maintaining synaptic integrity. Loss of Bassoon and Piccolo leads to the aberrant degradation of multiple presynaptic proteins, culminating in synapse degeneration. This phenotype is mediated in part by the E3 ubiquitin ligase Siah1, an interacting partner of Bassoon and Piccolo whose activity is negatively regulated by their conserved zinc finger domains. Our findings demonstrate a novel role for Bassoon and Piccolo as critical regulators of presynaptic ubiquitination and proteostasis.


Subject(s)
Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Presynaptic Terminals/metabolism , Proteolysis , Ubiquitination/physiology , Animals , Cytoskeletal Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , RNA Interference , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Zinc Fingers
4.
J Neurosci ; 31(40): 14250-63, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976510

ABSTRACT

Filamentous (F)-actin is a known regulator of the synaptic vesicle (SV) cycle, with roles in SV mobilization, fusion, and endocytosis. However, the molecular pathways that regulate its dynamic assembly within presynaptic boutons remain unclear. In this study, we have used shRNA-mediated knockdown to demonstrate that Piccolo, a multidomain protein of the active zone cytomatrix, is a key regulator of presynaptic F-actin assembly. Boutons lacking Piccolo exhibit enhanced activity-dependent Synapsin1a dispersion and SV exocytosis, and reduced F-actin polymerization and CaMKII recruitment. These phenotypes are rescued by stabilizing F-actin filaments and mimicked by knocking down Profilin2, another regulator of presynaptic F-actin assembly. Importantly, we find that mice with a targeted deletion of exon 14 from the Pclo gene, reported to lack >95% of Piccolo, continue to express multiple Piccolo isoforms. Furthermore, neurons cultured from these mice exhibit no defects in presynaptic F-actin assembly due to the expression of these isoforms at presynaptic boutons. These data reveal that Piccolo regulates neurotransmitter release by facilitating activity-dependent F-actin assembly and the dynamic recruitment of key signaling molecules into presynaptic boutons, and highlight the need for new genetic models with which to study Piccolo loss of function.


Subject(s)
Actins/chemistry , Actins/physiology , Cytoskeletal Proteins/physiology , Neuropeptides/physiology , Presynaptic Terminals/physiology , Protein Multimerization/physiology , Animals , Animals, Newborn , Cells, Cultured , Female , Gene Knockdown Techniques/methods , Male , Mice , Mice, 129 Strain , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...